Bale 2015

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

REVIEWS

STRESS

Epigenetic and transgenerational


reprogramming of brain development
Tracy L. Bale
Abstract | Neurodevelopmental programming — the implementation of the genetic and
epigenetic blueprints that guide and coordinate normal brain development — requires tight
regulation of transcriptional processes. During prenatal and postnatal time periods,
epigenetic processes fine-tune neurodevelopment towards an end product that determines
how an organism interacts with and responds to exposures and experiences throughout life.
Epigenetic processes also have the ability to reprogramme the epigenome in response to
environmental challenges, such as maternal stress, making the organism more or less
adaptive depending on the future challenges presented. Epigenetic marks generated within
germ cells as a result of environmental influences throughout life can also shape future
generations long before conception occurs.

Environmental changes that perturb an animal’s home- (that is, as an adult) may be more limited in their effect
ostasis — including stress, nutritional challenges and and duration, as they may produce specific epigenetic
infection — can influence the developing and matur- changes that do not manifest in epigenomic or germ cell
ing brain and thereby alter the original blueprint and reprogramming.
establish a new one in a process known as reprogram- According to the Barker hypothesis6, neurodevelop-
ming 1. There is a great history of studies that have mental reprogramming induces adaptations to the early
examined this process, and it has recently become clear environment in anticipation of a similar postnatal envi
that epigenetic mechanisms, including DNA meth- ronment. When there is a mismatch between these two
ylation, histone modification and the actions of small environments, such reprogramming may increase disease
non-coding RNAs, have an important role in such risk. For example, if a mother is exposed to increased
neuro­developmental reprogramming 2–5. However, the stress during pregnancy but the postpartum environ-
precise mechanisms by which exposure to environmen- ment is not stressful, the offspring’s brain may continue to
tal perturbations can influence long-term outcomes are respond inappropriately to stressors that are experienced
highly complex and depend on the type and the severity throughout life6.
of the exposure, whether it occurs during a window of Recent reviews in this area have focused on the spe-
vulnerability in the target circuit or system, the organ- cific developmental periods (including prenatal, postnatal
ismal sex and whether there are epigenetic changes in and pubertal periods) during which such reprogramming
germ cells (FIG. 1). occurs, the types of stimuli that are involved (including
The timing of an environmental exposure or experi- specific diets and exposure to infection or to stress) and
ence may determine whether the event has limited and specific epigenetic outcomes2–5,7–9. The goal of this Review
specific epigenetic effects or whether it more broadly is to integrate these points and highlight potential shared
affects the epigenome (BOX 1) as well as how long the mechanisms that are important in reprogramming the
effect persists (acutely or across generations). As epige- developing brain. I examine results from human and
Department of Biomedical
netic processes are integrally involved in neurodevel- animal studies that shed light on the factors that direct
Sciences, School of Veterinary
Medicine and Department of opment and maturation, insults that are experienced and shape brain development, and I discuss how differing
Psychiatry, Perelman School during these key periods (which roughly correspond parental contributions may affect offspring disease risk
of Medicine, University of to early or late gestation) would be predicted to repro- and resilience. In particular, I examine transgenerational
Pennsylvania, Philadelphia, gramme the epigenome more extensively and, if also epigenetic effects on neurodevelopment and discuss the
Pennsylvania 19104, USA.
e-mail: tbale@vet.upenn.edu
incorporated into the germ cells, their effects may interactions between specific perturbations, develop-
doi:10.1038/nrn3818 become transgenerational1. By contrast, events that mental windows of susceptibility and the sex-specificity
Published online 29 April 2015 occur outside a sensitive period of brain development of these effects.

332 | JUNE 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Environmental Maternal milieu Fetus The importance of the prenatal period. The best evidence
exposures • Cytokines Epigenetic programming of that the timing of insults is a factor in disease risk comes
• Maternal diet • Lipids germ cells and somatic
• Maternal stress • Stress hormones tissue: methyl donors (folate from birth cohort studies examining the effects of maternal
• Maternal infection • Metabolic indices and choline) broadly affect (prepartum) exposure to specific stress or immune chal-
(glucose, insulin the methylome or specific lenges. These studies compile birth registries that enable
and FFAs) imprinted genes
prospective studies to be carried out and thus may have
access to more accurate records of the exposure under
investigation than do retrospective studies. They also
allow the collection of plasma and tissue samples, which
enables potential biomarkers and genetic and/or epigenetic
factors associated with susceptibility to be assessed. For
example, birth cohort studies have revealed strong asso-
ciations between in utero exposure to infection, hypoxia
or starvation and an increased risk of schizophrenia22,23,
as well as an association with impairments in executive
function in patients with schizophrenia15, which indicates
that prenatal brain development, not surprisingly, is a
Placenta uniquely susceptible period. Furthermore, recent stud-
• Cytokines ies showed marked changes in cortical layer patterning,
• Nutrient transporters and receptors which is a prenatal process, in children with ASD, suggest-
(folate and choline)
• Lipid metabolites ing that a disturbance occurred before birth24. However,
• Growth factors (IGF and IGFBP) in‑depth neuro­developmental assessments that compare
• Oxygen perfusion similar perturbations across these prenatal and postnatal
• Glycogen
periods will be necessary to identify a specific period of
development as a crucial factor in disease risk.
Nature Reviews | Neuroscience
Figure 1 | Complex interactions between the maternal milieu, placenta and fetal There is growing evidence that there are specific devel-
compartments during gestation. Fetal antecedents such as maternal dietary opmental windows of increased vulnerability throughout
deficiencies, chronic stress or infection can promote endocrine disruptions in the pregnancy itself 10,25–27. Indeed, a recent epidemiological
maternal milieu, including increases in pro-inflammatory cytokines and stress hormones, study reported an association between maternal stress
and shifts in metabolic indices. In addition, these environmental exposures can indirectly that is experienced specifically during the first trimes-
alter placental development and function by changing local energy metabolism and lipid
ter of pregnancy and an increased risk of schizophrenia
storage and metabolism. This can alter the transmission of key nutrients that are
important for fetal growth and brain development, including growth factors and methyl in male offspring 28. Exposure to maternal infection and
donor nutrients such as folate and choline, which can affect fetal somatic and germ cell high levels of pro-inflammatory cytokines during early or
epigenetic programming. FFAs, free fatty acids; IGF, insulin-like growth factor; IGFBP, mid-gestation was also associated with an increased risk
IGF-binding protein. of neurodevelopmental disorders18. For example, children
of women who were exposed to influenza during early or
mid-pregnancy, but not during late gestation, had a three-
Developmental windows of susceptibility fold higher risk of schizophrenia than those of women
Studies evaluating susceptibility factors in neuro­ who had not been exposed13. Similarly, studies using the
developmental disorders have indicated that certain Danish medical birth register reported that maternal viral
outcomes are commonly linked to exposure to differ- infection resulting in hospitalization in the first trimester,
ent types of gestational and early-life environments. For but not later in pregnancy, was associated with a three-
Birth cohort studies example, exposure to stress or infection during either of fold higher risk of ASD in the child29,30. Furthermore, the
Longitudinal prospective these developmental periods is linked to an increased Early Markers for Autism study found increased levels of
studies in which pregnancies
risk of depression, schizophrenia and autism spectrum 17 cytokines in early and mid-gestation in the mothers
are followed through birth and
into childhood to correlate the disorders (ASDs)1,10–15. This suggests that these insults of children that were later diagnosed with ASD31. These
risk for given outcomes with may promote common mechanisms irrespective of the studies broadly suggest that early pregnancy is a particu-
specified experiences or time at which they occurred in development. However, larly sensitive period in development, during which time
exposures during gestation. epidemiological evidence also supports the idea that insults such as stress and inflammation can promote pro-
These cohorts of individuals
differ from other
the time of exposure during development may be gramming changes that will increase the risk of disease.
epidemiological studies in important for some types of environmental challenge, This increased vulnerability may lie in the sensitivity of
which the outcome measures as similar perturbations occurring at earlier or later the brain, the reactivity of the mother in early gestation
are mainly retrospective. stages have been associated with different long-term and/or the ability of the fetus to recover from such insults.
outcomes1,16–18. Prepubertal and pubertal brain matu- In addition to stress or infection during pregnancy,
Biomarkers
Specific biochemical, ration has been less well examined; however, these reduced maternal nutrition can also have a negative
molecular, anatomical or periods also seem to be susceptible to environmental influence on fetal growth and can induce epigenetic
physiological characteristics disturbances. This is especially true for the prefrontal changes, thereby influencing fetal brain development.
that are used to predict, cortex and its crucial connections within the limbic Long-term epidemiological studies investigating fam-
measure or indicate the
presence or progress of
system, in which disruptions have been linked to an ine exposures during specified periods of pregnancy or
disease or the effects of increased risk of attention deficit hyperactivity disorder postnatal life are beginning to discern associated risks for
treatment. (ADHD) and schizophrenia19–21. neuro­psychiatric disease in first- and second-generation

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | JUNE 2015 | 333

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Defining epigenetics and epigenomics


to a specific period of gestation or even to be limited to
the prenatal environment. However, maternal body mass
The definition of the term ‘epigenetic’ has been subject to continuous evolution. and obesity preceding and during pregnancy have been
Waddington172 coined the term to describe the premise that genes can respond to a linked to severity in childhood ADHD symptoms and
dynamic environment and thereby shape phenotypes such that they are acutely other executive function deficits36,37. Exposure to mater-
advantageous to the individual (as opposed to advantageous on a much longer
nal obesity during gestation has also been associated with
evolutionary scale). A more recent operational definition narrowed the usage to
include the necessity of heritability, either across generations or to daughter cells,
a twofold–threefold increased risk of schizophrenia in
which involves the erasure and subsequent replacement of epigenetic marks173. offspring in two separate birth cohorts32,33.
However, over the past decade, developmental and behavioural neuroscientists have As is clear from the findings described above, gesta-
used the term ‘epigenetic modifications’ to refer to functionally relevant chromatin tion is one of the most studied developmental windows
modifications that alter gene expression but that do not involve a change in DNA of vulnerability 38,39 and is a critical time in neuro­
sequence. Recent studies have produced mechanistic insight both at the level of developmental programming 13–15,24,32. In this context, it
epigenetics, which examines changes in the expression of specific genes or loci that is important to consider that early pregnancy involves
result from the addition of epigenetic marks, such as DNA methylation, or from processes such as embryo implantation and placental dif-
changes in the levels of a given microRNA, and at the level of epigenomics, which ferentiation, whereas mid- and late gestation are charac-
examines genome-wide changes in gene expression that can be attributed to broad
terized by somatic growth, including considerable brain
chromatin modifications. Such outcomes, which might be directed by a given histone
modification (such as histone H3 lysine 4 (H3K4) acetylation) or by changes in the
development 40. Therefore, exposure to environmental
methylome, can influence large sets of genes that are involved in specific functions challenges will influence different developmental pro-
and that generate changes in developmental programming in response to an insult or cesses depending on when in pregnancy the exposure
a perturbation9. occurred. In addition, the primordial germ cells, which
will give rise to the next generation, undergo remethyla-
tion of imprinted genes before birth in males, whereas
offspring. Therefore, these studies have the potential to this occurs postnatally in females40. Therefore, if an insult
uncover transgenerational epigenetic mechanisms as well occurs during a period in which the germ cells are under-
as direct effects on neurodevelopment. Initial studies have going this normal epigenetic process, changes could be
reported an increased risk of neurodevelopmental disor- incorporated that would give rise to a phenotype in future
ders in children of women who were exposed to severe generations.
Executive function caloric restriction or famine during pregnancy compared
A set of cognitive abilities that with other periods of postnatal development 32,33. For Mechanisms of reprogramming
include inhibition (resisting instance, children of women who were pregnant during Animal models have been used in an attempt to mimic
habits, temptations or the Dutch ‘hunger winter’ of 1944–1945 were more likely the epidemiological findings described above by exam-
distractions), switching
(adjusting to change) and
to have altered stress responsiveness and schizophrenia ining offspring outcomes that result from specific types
working memory (mentally as adults32,34. These outcomes were also associated with of insults during specific periods of development. These
holding and using information). changes in circulating growth factors and altered DNA studies have provided an insight into the mechanisms that
methylation of several genes, including the maternally are involved in reprogramming the brain. Rodent brain
Stress responsiveness
imprinted gene insulin-like growth factor 2 (IGF2), which development begins during gestation but mostly contin-
Behavioural or physiological
measures in response to a were measured 60 years after the famine exposure35. In ues postnatally. Therefore, using these animal models,
stress provocation. In all addition, their second-generation offspring had increased researchers have been able to examine offspring pheno-
mammals, the physiological adiposity and overall worse health as adults32. These types that are relevant to neurodevelopmental disorders
hypothalamic–pituitary– transgenerational outcomes suggest that the initial mater- by studying maternal exposure to stress, dietary chal-
adrenal stress axis is the
standard measure of stress
nal dietary restriction induced epigenetic marks in fetal lenges and infection both during pregnancy as well as in
experience. Behavioural germ cells and thus the first-generation offspring were the early postnatal period.
changes in response to stress able to transmit a phenotype without re‑exposure to the
or threat can also be examined insult. Of course, epidemiological studies are limited by Prenatal maternal stress. Studies in which the pregnant
as an indication of stress state.
an inability to control for the effects of additional mater- dam is exposed to perturbations that provoke activation
Imprinted gene nal or paternal lifetime experiences and/or interactions of her neuroendocrine stress system have shown that the
A gene expressed from only with the maternal milieu in subsequent generations. In effect of maternal stress during pregnancy on offspring
one allele in a manner that addition, it is important to note that famine experience outcomes, including on stress sensitivity, reward and
depends on the parent of is also an enormous physiological and psychological cognition, depends on the gestational timing of the expo-
origin and that is regulated by
DNA methylation.
stressor, and stress insults are also metabolic challenges. sure (as well as on the sex of the fetus)41–47. In many spe-
Therefore, it is not surprising that both maternal stress cies, including mice, rats, guinea pigs and non-human
Hypothalamic–pituitary– and maternal famine early in pregnancy produce similar primates, maternal stress increases offspring stress
adrenal stress axis neurodevelopmental disorder risk13–15,32. sensitivity such that the overall production of gluco­
(HPA stress axis). The
The current obesity epidemic in Western society has corticoids following hypothalamic–pituitary–adrenal stress
neuroendocrine core of the
stress system. Its activation shifted the focus from understanding the effect of under- axis (HPA stress axis) activation is increased, behav-
results in the release of cortico- nutrition to examining the effects of maternal overnutri- ioural stress responses are heightened and cognitive
tropin-releasing factor from the tion on neurodevelopment. Indeed, it has been proposed deficits are present, all of which are endophenotypes
hypothalamus, adrenocortico- that transgenerational epigenetic mechanisms may con- that are associated with neuropsychiatric disease42,46,48–56.
tropic hormone from the
pituitary and cortisol
tribute to the rapid rise in obesity rates1. Maternal diet To compare the effects of maternal stress experi-
(corticosterone in rats and and body composition are less dynamic factors than stress ence across early, mid- or late pregnancy on the pro-
mice) from the adrenal glands. or infection and are therefore more difficult to designate gramming of offspring stress outcomes, studies in

334 | JUNE 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

mice and guinea pigs have used a chronic stress para- Postnatal maternal stress. Complex interactions between
digm42,46,49,50. In mice, exposure to stress very early in the neonate and its mother occur in the postnatal period,
pregnancy (embryonic days 1–7) resulted in male, but and maternal behaviours such as nursing and grooming
not female, offspring that showed increased immo- can strongly influence neurodevelopment 63–77. Rodent
bility in both tail suspension and forced swim tests studies have shown that both enhanced and fragmented
as well as cognitive deficits in spatial learning and maternal care in the early postnatal period can result in
memory tasks as adults42,50. Early stress exposure also marked differences in the behaviours of the adult off-
affected the programming of stress neurocircuitry: spring. It is well established that the quality of maternal
male offspring showed increased corticosterone pro- care influences epigenetic processes in the developing
duction in response to acute restraint stress, and this brain8,73,78. This was first shown in a rat model in which
was associated with increased levels of corticotropin- dams showed consistent high or low levels of maternal
releasing factor (CRF) in the amygdala and reduced care — identified as increased or decreased licking and
hippocampal gluco­c orticoid receptor expression 42. grooming — and this has since been demonstrated in
These changes were associated with corresponding many additional species, including mice, hamsters and
alterations in promoter methylation, which suggests non-human primates65–67,70,73,76,79–82. These studies have
that the prenatal stress experience resulted in long- shown that the mother’s interactions with her offspring
term epigenetic reprogramming of genes involved in predict their behavioural and hormonal responses to
the development of stress neurocircuitry. The timing stress in adulthood. Specifically, in rats, high rates of
specificity of the effects of prenatal stress was simi- licking and grooming by the mother were associated
larly explored using short periods of daily flashing with more rapid negative feedback to the HPA stress axis
light as a maternal stressor in guinea pigs at mid- or as a result of enhanced glucocorticoid receptor sensitiv-
late gestation49. Adult offspring that were exposed to ity as well as reduced levels of anxiety-like behaviours72.
stress in mid-gestation showed increased anxiety-like At an epigenetic level, these outcomes were correlated
behaviours, increased basal corticosterone levels and with changes in DNA methylation of the glucocorti-
lower hippocampal gluco­corticoid receptor expres- coid receptor promoter and in glucocorticoid receptor
sion; however, late-gestation stress did not produce expression in the hippocampus74. Although other genes
these outcomes. This evidence for specific effects of are probably also affected, the glucocorticoid receptor
maternal stress during mid-gestation complements gene, Nr3c1, seems to have been the most consistently
the evidence arising from the epidemiological studies examined across models and studies. NR3C1 expression
described above28,47,49. and promoter methylation has also been examined in
Interestingly, in many studies, male offspring are humans following early-life trauma, showing a poten-
the most severely affected sex. They often present with tially translational outcome for an epigenetic finding
a ‘dysmasculinized’ phenotype; that is, the prenatally from animal models83,84. However, to adequately address
stressed males seem to be more similar to control the question of how maternal stress during pregnancy
females than to control males in terms of their behav- ultimately reprogrammes the developing brain, it is nec-
iours, physiology and gene expression patterns. For essary to consider the epigenome and the broader epige-
instance, male mice exposed to maternal stress very netic mechanisms that would be capable of establishing
early in gestation showed significant changes in spatial changes that are marked enough to alter organismal
learning and memory performance in a Barnes maze and behaviour decades later.
Endophenotypes used ‘female-like’ strategies to solve the task50. In addi- Chronic stress during the postnatal period — affecting
Quantifiable phenotypes with
an assumed intermediate role
tion, these mice showed dysmasculinized patterns of the dam and/or the pups — can also affect the cognitive
in the pathway from genes to stress physiology, behaviour and cognitive performance, performance of adult offspring through functional and
complex phenotypes. It is which indicates a potential disruption in normal perina- structural changes in specific brain regions, including
thought that the action of an tal brain masculinization44,52. Similar dysmasculinizing the hippocampus and the hypothalamus. For example,
endophenotype is easier to
results have been reported in prenatally stressed rats and in a rat model of altered early postpartum maternal care
understand biologically and
genetically than the action of guinea pigs43,46,47,49,57,58. Interestingly, several recent stud- resulting from the stress of repeated maternal separation,
the complex phenotype of ies have reported an association between steroidogenic the offspring showed lifelong epigenetic changes in the
primary interest. They enable activity and testosterone levels in fetuses and autism or hippocampus68. Specifically, the ability of a transcrip-
the examination of specific autistic-like traits in young children59, and low serum tional repressor to bind to Crf was decreased because of
aspects of complex human
diseases in animal models.
testosterone levels have been associated with certain increased DNA methylation, and this was associated with
schizophrenia symptoms such as reduced verbal and a reduced ability of the offspring to appropriately respond
Barnes maze working memory in males60. Fetal testosterone levels to and to cope during stressful experiences as adults. This
A spatial learning and memory also predict the area of the sexually dimorphic grey mat- shows an interesting epigenetic reprogramming effect in
task in which the animal is
ter in the human brain61, and male patients with schizo- response to postnatal stress that opposes changes that
placed on a large, open table
that has holes around the phrenia have orbitofrontal cortex-to-amygdala volume have been observed following prenatal stress: that is, the
circumference. Only one of the ratios that are more similar to those of control women Crf promoter was hypomethylated following postna-
holes has an escape tunnel, than to those of control men62. These findings support tal stress42. This again highlights the importance of the
and the animal must learn to the idea that brain masculinization is an important timing of environmental exposures in producing their
use distal cues to identify this
hole and escape in the shortest
part of normal brain development in males and sug- long-term consequences. Interestingly, similar long-term
possible time. Learning in this gest that this process may be a point of vulnerability to changes in the expression of CRF in the hypothalamus
task involves the hippocampus. perturbations in the environment. were also found in offspring in a model of fragmented

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | JUNE 2015 | 335

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

maternal care, in which the rat dam is given limited and The placenta may function as an important interme-
insufficient material with which to build her nest, sug- diary between an unhealthy maternal milieu and fetal
gesting that alterations in maternal behaviour during markers of neuroinflammation by producing its own
early postnatal life can drive similar epigenetic events in pro-inflammatory cytokines. Placentas from pregnant
her offspring 65,70. macaques receiving a high-fat diet throughout preg-
nancy showed increased levels of the pro-inflammatory
Maternal nutrition and obesity. Rodent studies have cytokines tumour necrosis factor (TNF) and inter-
shown a link between prenatal availability of nutrients leukin‑6 (IL‑6), and of Toll-like receptor 4 (REF. 102).
and offspring brain development 85. In mice, females Surprisingly, in pregnant rats with restricted access
receiving a protein-deficient diet throughout preg- to low-protein food, levels of TNF and IL‑6 were also
nancy and lactation produced offspring with intrauter- increased in placental tissue103. Timing also seems to be
ine growth restriction and marked changes in dopamine important for the interaction between the immune sys-
circuitry and reward-related behaviours as adults86. tem and the placenta, as maternal immune activation
Interestingly, the dopamine reward neurocircuitry was during early gestation in mice can result in implanta-
also affected when pregnant dams were fed a high-fat tion failure104,105, and mid-gestation inflammation in
diet throughout pregnancy and lactation, which suggests the placenta has been linked to schizophrenia-like and
that the developing reward system is particularly sensi- ASD-like phenotypes106.
tive to gestational nutrition (probably stemming from
an evolutionary benefit of linking motivational behav- Maternal infection and immune activation. Animal
iours with caloric need)87–92. Reduced reward sensitivity studies examining the effects of maternal infection
(anhedonia) is a symptom of depression in humans; thus, during pregnancy have reported many behavioural
there may be a connection between prenatal dietary per- outcomes in the adult offspring that are similar to
turbations and an increased risk of affective disorders or those detected in offspring exposed to prenatal stress,
addictions. As described above, maternal undernutrition including increased stress reactivity and impaired cog-
also increases stress responsivity in pregnant females and nitive performance11,12,18,107. This suggests that the two
thus contributes to offspring phenotypes that are simi- manipulations may affect the fetus and the developing
lar to those produced by maternal stress93. For example, brain through common mediators, including glucocor-
rats that were exposed to 50% food restriction during ticoids and/or pro-inflammatory cytokines96,108–112. In
pregnancy had increased plasma corticosterone levels fact, inhibition of inflammatory processes in a mouse
and their adult offspring had higher adrenal weights94. model of maternal chronic stress ameliorated the effects
These maternally food-restricted offspring also showed of stress on the offspring 108. The pro-inflammatory
changes in the expression of key stress genes, includ- cytokines produced by the mother and/or in the placenta
ing reduced levels of hippocampal Nr3c1 expression. during maternal infection or chronic stress can elicit
Furthermore, exposure of female guinea pigs to nutri- their long-term effects on fetal somatic cells through
ent restriction for 2 days in the last third of pregnancy interactions with epigenetic machinery. For example,
increased maternal and fetal cortisol levels and reduced IL‑6 increases the expression and the activity of DNA
the expression of glucocorticoid receptor in the offspring (cytosine 5)-methyltransferase 1 by regulating its gene
hypothalamus and hippocampus in adulthood95. These promoter 113. In another example, in a mouse model of
outcomes are similar to those reported in studies inves- early pregnancy stress, changes in the expression of the
tigating the effects of prenatal stress and maternal dexa- chromatin-remodelling enzyme O‑GlcNAc transferase
methasone treatment 1,22,42,55,56,94,96–98, which suggests that (OGT) were detected in male but not in female placental
common signals are produced in the maternal milieu tissue and were found to regulate a broad pattern of gene
during maternal stress and undernutrition. These com- expression that is associated with the active histone mark
mon signals are probably related to the similar catabolic H3K4me3 (histone H3 lysine 4 trimethylation)38,114.
metabolism that occurs during these experiences93.
Maternal dietary challenges also affect the maternal Transgenerational programming
and fetal immune systems. For instance, increases in the Although some environmental factors only ‘programme’
number of hypothalamic microglia and pro-inflamma- the individual that is directly exposed to them, others
tory cytokine levels were found in fetuses of macaque are transmitted across one or more generations115. The
mothers who received a high-fat diet before and during transmission of epigenetic marks to future generations
pregnancy 99. As juveniles, the female, but not the male, through germ cell reprogramming adds complexity to
offspring showed increased anxiety-like behaviours100. our attempts to establish causal links between environ-
Similarly, increased numbers of activated microglia in mental risk factors and disease. True transgenerational
the hippocampus were reported for the adult offspring reprogramming requires an epigenetic mark to be erased
of rat dams fed a diet high in saturated fat throughout and then re‑established in the germ cell in the absence
pregnancy and lactation and these animals also showed of re‑exposure or continuation of the exposure115 (FIG. 2).
increased anxiety-like behaviours and deficits in spa- Tight control of epigenetic regulation is crucial during
tial learning 101. Together, these studies indicate that the early phase of gestation, which is characterized by
Microglia
Glia of mesodermal origin and
there are sex-specific interactions between the mater- a wave of genome demethylation followed by de novo
the resident macrophages of nal diet during early life and long-term neuroimmune remethylation, establishment of imprints and sex deter-
the CNS. programming 100,101. mination, and is therefore particularly sensitive to

336 | JUNE 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

F1 F2 F3 mothers are stressed during pregnancy), which results


in increased stress sensitivity and an increased cortico­
a Terminal somatic
sterone response to acute restraint stress as adults, pass
programming in utero
on this phenotype to their male, but not their female,
b Somatic and primordial offspring 119. Similarly, when mice were exposed to early
germ cell programming
in utero postnatal stress (chronic and unpredictable maternal
separation from postnatal days 1 to 14), their stress-
c Stable germline
inheritance sensitive behavioural phenotype in adulthood contin-
ued through to the third generation120. Changes were
d Transgenerational
perpetuation of detected in DNA methylation in the germ line of this
maternal exposure third generation, which suggests that germ cell epi­
e Transgenerational genetic marks can be maintained across generations
perpetuation of without re‑exposure to the insult 120. These studies sup-
paternal exposure port the idea that the germ line can be manipulated, and
they show the potential ability of external influences to
Shows phenotype No phenotype increase susceptibility or resilience to disease in subse-
quent generations. It is important to note that an envi-
Nature Reviews | Neuroscience
Figure 2 | Modes of maternal and paternal transgenerational epigenetic ronmental signal that can influence the germ line does
transmission. Several mechanisms for transgenerational epigenetic transmission are not necessarily produce a notable phenotype in the first
possible, as shown in the table, in which a red mouse indicates that the phenotype is generation if there is no effect on somatic tissues (includ-
shown in a particular generation. a | In utero somatic programming may affect tissue and ing the brain). Whether such an effect occurs probably
brain development in the fetus, initiating a developmental trajectory that results in an depends on the timing, the type or the severity of the
adult phenotype in the first (F1) generation. However, this phenotype is not transmitted exposure121. However, in such a scenario, if the germ cell
to the next generation, as there is no germ cell involvement. b | If in utero somatic were reprogrammed this would give rise to a phenotype
programming is accompanied by changes in primordial germ cells, the phenotype will be in the second generation; thus, phenotypes can skip a
transmitted to a second (F2) generation. In order for this to occur, germ cells must be generation, making it more difficult to form clear risk
exposed to a maternal insult during gestation. c | If the in utero exposure is able to alter
analyses for pregnant women or for other at‑risk groups
epigenetic marks in the germ line of the F1 offspring and this reprogramming is able to
(FIG. 3). As this premise has not yet been mechanistically
withstand erasure and re‑establishment during subsequent generations’ germ cell
development, the phenotype will be perpetuated across generations without the need examined, the specific signals or insults remain to be
for re‑exposure to the original insult. This is defined as a truly transgenerational determined.
epigenetic programming. d | Transgenerational phenotypes may also occur through a
repeated maternal effect by which programming during gestation or through exposure Maternal transmission. Studies in which female mice
to altered maternal behaviours results in an adult phenotype that is only found in the were exposed to nutritional challenges have provided
maternal lineage. In order for this to occur, the offspring must be re‑exposed to the insult some of the clearest examples of how phenotypes can be
in each generation to promote the continuation of the phenotype from the dam. This lost or retained across generations121–126. These studies
trait would not be passed through a paternal lineage. e | Transgenerational programming have monitored body length, adiposity, glucose regu-
through a paternal lineage originating from an initial exposure in the male requires
lation, metabolic rate and behaviour through at least
establishment or reprogramming of epigenetic marks in the sperm. Adapted with
permission from REF. 115, Elsevier. three generations and have reported epigenetic changes,
including effects on imprinted genes. Maternal obe-
sity or exposure to a high-fat diet during pregnancy is
maternal influences that could result in embryonic and associated with reduced insulin and glucose sensitiv-
germ cell reprogramming 40. The different ways in which ity and increased body size and adiposity in the first-
germ cell epigenetic marks are transmitted and inherited generation offspring of humans, non-human primates
can also be examined; these include whether the mother and rodents122,127–131. However, not all of these traits will
and/or the father pass on the trait and whether male or arise in the second generation, which supports the idea
female offspring (or both) inherit it. Epidemiological that many phenotypes are the result of direct somatic cell
data from Swedish famine periods suggest that there may programming and not of altered epigenetic marks in the
be specific developmental windows during which germ germ cell. For example, one study in mice found that an
cells are more vulnerable to this type of environmental increased offspring body size was the only phenotype of
influence116; boys exposed to famine before puberty were many from the first generation that was retained into the
more likely to have children and grandchildren with an second and the third generations following the initial
increased risk of cardiovascular disease and diabetes exposure to a maternal high-fat diet during pregnancy 123.
than males exposed outside this period117. However, A closer examination of the transmission pattern of this
studies have also shown that the type of perturbation or phenotype revealed that only second-generation fathers
exposure may be an important determinant in how or if could pass on the trait and only to their third-generation
the germ cells are affected118. daughters, indicating an epigenetic mechanism (as male
Animal models have been used to examine the mice were not permitted any interaction with the litter,
transgenerational outcomes of germ cell reprogram- any passage of traits to their offspring must have been
ming that are induced by maternal and/or paternal programmed in their germ cells) involving a pater-
exposure to an environmental factor. For example, male nally expressed gene (as only the second-generation
mice that are exposed to stress in utero (that is, their fathers were able to pass on the trait) that may be on the

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | JUNE 2015 | 337

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Pregnant female (F0) which to study mechanisms of epigenetic transmis-


sion than models of maternal transmission, which are
Somatic tissue complicated by the potentially confounding effects
(including brain) of maternal behaviours, parturition and lactation. As
Placental stated above, the term ‘transgenerational’ requires that
contribution an epigenetic mark giving rise to a phenotype is found
in the germ cell and is passed on to the next generation
in the absence of re‑exposure. Therefore, to be consid-
ered a transgenerational epigenetic mechanism in a male
Maternal Fetus (F1) model of transmission, a phenotype need be detected
stress only into the second generation. Perturbations that
and diet
occur during pregnancy, such as maternal stress or infec-
tion, must affect the third generation for their influence
to be truly transgenerational, because the gestating first-
Primordial
germ cell (F2) generation offspring and the germ cells that will give rise
to the second-generation offspring are simultaneously
exposed to the initial perturbation115 (FIG. 2).
Epigenetic marks normally found in sperm include
changes in the expression of non-coding RNAs (micro-
RNAs (mi­RNAs) and PIWI-interacting RNAs (piRNAs;
which are important in silencing retrotransposons)),
histone modifications (for the few retained histones in
sperm) and DNA methylation140. Perturbations such
as stress can alter these marks, resulting in offspring
Figure 3 | Programming of phenotypes and disease risk can skip generations.
Maternal stress or dietary insults can generate signals, in the form of changes in the with detectable phenotypes. For instance, when male
hormonal milieu, that are transmitted from the maternalNature Reviews
to the fetal | Neuroscience
compartment via the mice were exposed to chronic social defeat or chronic
placenta. The resulting maternal programming of fetal somatic tissues can lead to variable stress before breeding, their offspring showed
changes in long-term health outcomes in the first generation (F1). Furthermore, in utero behavioural and physiological phenotypes of stress
exposure can also occur for the primordial germ cells that will give rise to the second dysregulation, including altered HPA axis responses
generation (F2), as these germ cells are also present and undergo reprogramming during to stress132,134. Furthermore, one study showed that,
embryonic development. If the exposure does not produce a detectable effect on in sperm from fathers who were chronically stressed
somatic tissues but is still able to reprogramme epigenetic marks in the germ cell, a either during puberty or as adults, the levels of a sub-
phenotype could effectively arise in the F2 generation that was not present in the F1
set of specific mi­RNAs were increased134. Interestingly,
generation, thus having the appearance of ‘skipping a generation’. Adapted with
rewarding or reinforcing environmental factors, such as
permission from REF. 115, Elsevier.
drugs of abuse, pleasant odours or high-fat diets, can
also produce germline epigenetic marks that are trans-
X chromosome (as only their daughters inherited the mitted to the offspring. For example, male rats exposed
trait)123. Indeed, the expression pattern of imprinted genes to a specific odour paired with a shock passed this
from these females supported this hypothesis: paternally conditioned experience to their offspring through an
expressed genes showed increased overall expression in epigenetic germ cell mechanism133. Similarly, male off-
the livers of third-generation female offspring born to the spring of male rats that were exposed to chronic cocaine
high-fat diet lineage second-generation males but not in self-administration showed reward neurocircuitry
those born to the females. hypofunction136. This phenotype was associated with a
change in histone H3 acetylation at the brain-derived
Paternal transmission. Although transgenerational neurotrophic factor (Bdnf) promoter in the sperm of the
effects of various changes in the maternal environment sires that were initially exposed, and the same change
are well characterized, fewer studies have examined pos- was also detected in the brains of their male offspring.
sible paternal modes of transmission. In such studies, In another study, female but not male offspring of male
male mice or rats exposed to social defeat, chronic stress, rats that were exposed to a high-fat diet for 10 weeks
cocaine, a high-fat diet, endocrine disrupters or olfactory had reduced glucose sensitivity and β-cell dysfunction135.
Social defeat cues before breeding have been examined132–139. These Such sex-specific outcomes suggest that the epigenetic
A behavioural model in which models provide unique opportunities to examine mech- mark or machinery involved is found on the X or the
rodents (predominantly males) anisms of transgenerational transmission. In a research Y chromosome and/or that the offspring phenotype is
are repeatedly exposed to a
laboratory environment, male rodents do not normally dependent on programming or activational effects of
more aggressive conspecific.
The outcomes of these participate in the rearing of their offspring and thus have oestradiol or testosterone. The mechanism by which
confrontations are repeated no postpartum behavioural influence on their develop- paternal life experiences such as stress, diets or drugs can
losses for the subject. This ment. Therefore, the ability of the sire to transmit a trait reprogramme epigenetic marks in the germ cell are not
exposure is then followed by a to his offspring is limited to the genetic and the epige- currently understood; however, normal processes and
period of housing the defeated
animal in close proximity to the
netic material in his sperm, which is an accessible and development across spermatogenesis and the stages at
aggressor to establish the plentiful tissue source for epigenetic analyses. Therefore, which epigenetic or transcriptional machinery is present
conditioning. paternal models provide a more transparent method by in the cell must be considered (FIG. 4).

338 | JUNE 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Epididymis

Window of vulnerability for programming and reprogramming epigenetic marks during spermatogenesis

Active transcription Meiotic Active transcription Chromatin


divisions compaction

RNA storage, RNA-binding proteins and translational repression


Chromatid body

Spermatogonium Early Late 1st meiotic 2nd Round Step 8 round Elongating Final sperm
spermatocyte spermatocyte division spermatocyte spermatid spermatid spermatid maturation

Figure 4 | Windows of vulnerability to environmental reprogramming in spermatogenesis. Nature The figure| Neuroscience
Reviews shows the
stages of spermatogenesis and sperm maturation, highlighting the periods in which environmental exposures can
reprogramme epigenetic marks. Active transcription and storage of RNA is ongoing through the spermatogonium,
spermatocyte and spermatid stages. However, once compaction of paternal DNA has occurred, which involves replacing
the majority of histones with protamines, there is not a clear mechanism by which epigenetic marks can be further altered
in the mature sperm. This suggests that an environmental exposure must occur within a sensitive time period of
spermatogenesis in order to pass on a new or reprogrammed epigenetic mark to future offspring. In most mammals,
complete spermatogenesis occurs in 6–8 weeks, by which point sperm are present in the epididymis for final maturation
processes. The time between chromatin compaction and the transit of mature sperm to the epididymis is at least 10 days,
during which time epigenetic and transcriptional machineries are no longer active. However, the secretion of exosomes
and other factors that interact with the maturing sperm in the epididymis may impart lasting epigenetic marks, such as
microRNAs. Figure is reproduced from Bale T. Lifetime stress experience: transgenerational epigenetics and germ cell
programming. Dialogues Clin. Neurosci. 2014;16:297–305 (REF. 179), with the permission of © AICH-Servier Research
Group, Suresnes, France, and adapted from REF. 180, Nature Publishing Group.

Sex specificity in epigenetic mechanisms There is also evidence that the effects of activation
As described above, epigenetic responses to environmen- of the maternal immune system on the offspring are
tal changes can be sex specific at several levels. The per- sex dependent. For example, male offspring of preg-
ception or the response to the exposure itself can differ nant rats exposed to lipopolysaccharides late in gesta-
between sexes, so that one sex produces a higher or a lower tion showed deficits in pre-pulse inhibition in adulthood,
change in hormone levels or greater neural activation, whereas female offspring were unaffected146. Similarly,
leading to a cascade of differing outcomes. The placenta male, but not female, mice exposed during late gestation
is an example of a tissue that shows sex-specific effects; to maternal injection of the viral mimic polyinosinic–
for example, maternal stress or dietary challenge during polycytidylic acid (poly(I:C)) showed behavioural and
pregnancy produces lasting gene expression changes in cognitive inflexibility in adulthood, resembling some of
the placentas of male fetuses but not in the placentas of the negative symptoms of schizophrenia18. These effects
female fetuses38,39,42. This provides a mechanism by which were also associated with reduced levels of glutamate in
the effects of stress can be transmitted in a sex-specific the prefrontal cortex in males. Such studies support the
manner to the developing brain. Epidemiological stud- widely held view that males are more vulnerable to pre-
ies have shown that gender is an important determinant natal insults than females, who are more susceptible to
in the severity and the onset of diseases that are associ- insults that occur in the postnatal environment 147.
ated with certain fetal antecedents. For example, male, Animal studies have also begun to examine the
but not female, children of women who were in their developmental timing of such sex-specific outcomes
second trimester of pregnancy during the 1940 invasion more closely1. For example, as described above, gesta-
of the Netherlands (and therefore who presumably suf- tion has been established as a period in which important
fered high stress levels at that time) had an increased risk masculinization of the male brain can be disrupted by
of schizophrenia141. A recent report found that maternal maternal stress22,42,50,57. Disruptions in the organizational
depression during pregnancy correlated with increased programming of the sexually dimorphic brain may also
postnatal anxiety development in children and that males have a role in the aetiology of neurodevelopmental dis-
Pre-pulse inhibition
were significantly more likely to be affected142. Many neu- orders. For example, structural brain volume analyses
A reduction in the magnitude
of the startle reflex that occurs rodevelopmental disorders, such as ASD, have a strong showed a reduction of the normal sexual dimorphism of
when an organism is presented sex bias143. Exposure to maternal stress before week 32 of the brain in male patients with schizophrenia62,148. These
with a non-startling stimulus (a gestation has been suggested to be a contributing factor studies are exciting because they suggest a specific time
pre-pulse) before being to ASD144. Therefore, studies examining the role of fetal window in development (probably prenatal) in which
presented with the startling
stimulus. Deficits in pre-pulse
sex in determining the specific outcomes of maternal an exposure or insult disrupted this normal masculin-
inhibition have been observed stress during pregnancy may provide a unique insight izing process. As the female brain is not exposed to
in patients with schizophrenia. into neurodevelopmental disease aetiologies145. gonadal hormones during this early developmental

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | JUNE 2015 | 339

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 2 | Common cellular mechanisms?


In thinking about the mechanisms by which prenatal perturbations promote transgenerational outcomes, it is interesting
to note that several seemingly different insults, such as maternal stress, diet and infection, can produce similar
neurodevelopmental and phenotypic outcomes. Mechanistically, this suggests that there are a limited number of
physiological signals from the maternal milieu that are responsible for transmitting information from the in utero
environment to the developing fetus and that physiological disturbances produced by maternal stress, infection, famine
or obesity share some common end points.
The most evolutionarily important signals are likely to be related to energy availability, which is important in
determining future fitness, and are therefore likely to include metabolic signals such as insulin, glucose, free fatty acids
and glucocorticoids, as well as inflammatory cytokines (FIG. 1). Glucocorticoids are certainly an important downstream
indicator of stress, and some aspects of prenatal stress models can be recapitulated by simply administering high levels of
glucocorticoids to the pregnant animal55,56,96,174,175. However, the stress response encompasses other stress hormones,
such as catecholamines, as well as numerous downstream metabolic factors. The involvement of these additional
metabolic processes in developmental programming at the level of the placenta and the fetus is probable and has been
shown to be important in rodent models of prenatal stress38,93,97,114,176.
There can only be a limited number of biochemical inputs that a given cell can to respond to and, as such, it is likely that
different environmental challenges impinge on the same signalling cascades and downstream effectors to promote
changes in existing epigenetic marks or to apply new ones — effectively reprogramming similar offspring phenotypes
from different starting points. Therefore, there must be a cellular barometer by which the quantitative and the qualitative
changes in the hormonal milieu are surveyed. For instance, during maternal or paternal stress exposure, for a signal to be
gauged as important enough to warrant activation of the epigenetic machinery to facilitate the creation of new or
changed epigenetic marks, does a stressor need to be of a certain duration, to reach a certain level (perceived as mild
versus severe or chronic) or to induce a certain level of stress hormones? How does a germ cell know whether a stressor
warrants passing on the information to future generations? Coincident detection of key signals occurring over precise
periods is probably required.

period, these findings indicate a sex-specific process Importantly, these regulatory mechanisms can be sensi-
that is a point of potential disruption in males. This tive to perturbations in the environment. As described
may ultimately lead to a mismatch for these males later above, maternal stress and infection are both associated
in life, as they would be genetically XY but would not with dysmasculinized phenotypes, which suggests that
have a fully masculinized brain. they cause a disruption in male sexual differentiation
Gonadal hormones and their receptors also interact or gonad development that in turn alters testosterone
with epigenetic mechanisms to promote normal brain production or its actions in the brain during this critical
maturation and development149. For instance, oestradiol neonatal organizational period1,155,156. Finally, it is also
can affect epigenetic machinery, including DNA methyl­ interesting to note that although the Y chromosome con-
transferases and histone deacetylases (HDACs), in the tains no miRNA genes, the mammalian X chromosome
neonate as a part of sexually dimorphic brain develop- is highly enriched for them and their density is approxi-
ment, and can alter DNA methylation patterns3,150–152. mately twofold higher than it is on autosomes in mice
Therefore, alterations in the levels of oestrogen, or its and humans147. In circumstances in which these genes
precursor testosterone, that result from perturbations escape X inactivation, females would have twice the
experienced during early life can manifest as long-term expression level of males, thus producing a sex-specific
reprogramming of brain function149. Oestradiol can also post-transcriptional mode of regulation that provides
alter histone modification through actions on HDACs149. females with the select ability to buffer or to respond
A single postnatal administration of an HDAC inhibitor differently to insults.
in male mice was shown to disrupt the sexual dimorphic
development of the bed nucleus of the stria terminalis Conclusions and future directions
(BNST), which is a crucial part of the neurocircuitry It is clear that the outcomes and the long-term sustain-
involved in regulating stress responses153. Small non- ability of dynamic epigenetic marks are dependent on
coding mi­RNAs also seem to be responsive to oestradiol factors such as the timing and the type of exposure,
levels in the early postnatal mouse brain; more than 250 the sex of the offspring and, in the case of transgenera-
of the most abundantly expressed mi­RNAs in the brain tional effects, the sex of the parent and the offspring.
significantly differ in their expression between males According to the Barker hypothesis, whether such
and females119. These sex differences are dependent on programming ultimately results in disease risk or resil-
the conversion of gonadal testosterone to oestradiol in ience mainly depends on the concordance between the
males154, as a single administration of an aromatase inhibi- expected and actual environments. As discussed above,
tor at birth was able to completely shift the entire male there are several common signalling mechanisms
miRNA expression pattern to that of a female119. through which perturbations during early develop-
These studies show that gonadal hormones can, ment are likely to function, including inflammation and
during important periods, promote epigenomic effects energy metabolism in the maternal milieu and in the
and thereby broadly influence neurodevelopment, placenta (BOX 2). Studies examining the early postnatal
effectively producing the sexually dimorphic brain. environment have also identified common epigenetic

340 | JUNE 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | Caution in translation of epigenetic outcomes


and ultimate brain function. Studies in mice focus-
ing on brain region-specific epigenetic processes and
We currently lack the ability to accurately predict who will present with a important behaviours related to cognitive performance,
neuropsychiatric disease. However, the collection of biological samples as part of learning and memory, and appetitive behaviours have
prospective studies may enable us to identify predictive biomarkers, which may include repeatedly shown how dynamic the epigenome contin-
epigenetic marks, and to distinguish between disease cause and effect if changes in a
ues to be into adulthood1,9,69,160–170. This reminds us that
given outcome precede disease onset rather than being a response to it. Of course, this
raises the question of whether epigenetic and epigenomic changes in peripheral cells
insults that occur early in life may be just a ‘first hit,’
and tissues are informative about what goes on in the brain, as most epigenetic changes which primes the brain to be more or less responsive to
are cell and tissue specific177. A great deal more evidence is still needed before we can future insults, with disease presentation occurring fol-
draw firm conclusions as to how accurate the epigenome will be in relaying the story of lowing multiple insults in life. Support for this hypoth-
an individual’s life exposures and experiences and our ability to translate that into esis can be seen in neuropsychiatric diseases in which
predictions for disease risk and resilience. adult stress-induced onset occurs, such as depression
Using established epidemiological and clinical measures to design better animal or post-traumatic stress disorder. Perturbations experi-
models that reproduce specific endophenotypes rather than attempting to model enced during neurodevelopment that promote dysregu-
mental health disorders in totality is an important goal that may facilitate study of the lation of stress pathways would certainly be considered
translational potential of the epigenome. Such models may enable us, for example, to
an important first hit 12,22,171.
better understand how an environmental factor may programme susceptibility or
resilience to disease in humans. However, if the field of transgenerational epigenetics
The field of transgenerational epigenetics has been
has taught us anything, it is that experience is everything. Results from studies showing on a fast-paced trajectory over the past decade, with
transgenerational effects of stress, odorant exposure, diet and infection have opened the development of animal models that reliably and
our eyes to the dynamic (that is, programmable and reprogrammable) epigenome, predictively mimic epidemiological studies in order to
including — and importantly — the germline epigenome5,133–135,178. The seemingly determine the mechanisms by which perturbations in
standard experiences that our experimental animals encounter, including shipping, the environment are associated with risk of neurode-
dietary changes, parasitic infections and treatments, construction noise and different velopmental disorders (BOX 3). Future studies will need
environments (such as germ-free versus conventional facilities), can produce to use ‘omic’ technologies to better understand the
substantial epigenetic differences within otherwise genetically identical inbred strains. broader reprogramming abilities of the environment
If present in the germ cells, these marks can last for generations, promoting phenotypic
on the epigenome. For instance, metabolomic analyses
variability within and between research laboratories. Controlling for such variables is
difficult but important.
of the maternal and the fetal milieu would be useful in
models of maternal stress or infection to determine the
metabolic signals that are involved in neurodevelop-
gene targets in the brain. In addition, recent studies have ment. There remain several considerable gaps in our
provided insight into the novel contributions of the gut knowledge: what are the physiological and the cellular
microbiome to early brain development; thus, metabolic signals by which experience is able to reprogramme
and immune processes may interact to ultimately affect an epigenetic mark (BOX 1), and when and how are the
neurodevelopment 157–159. Taken together with other germ cells involved? Identifying these mechanisms in a
important gene targets, these findings will provide the stepwise manner will facilitate the discovery of better
clues necessary to discover the upstream transcriptional disease predictive biomarkers and will perhaps lead to
regulatory mechanisms, which will increase our under- effective interventions for disease prevention and earlier
standing of the links between developmental experience treatments (BOX 3).

1. Bale, T. L. et al. Early life programming and 9. Sweatt, J. D. The emerging field of neuroepigenetics. 18. Meyer, U., Feldon, J. & Dammann, O. Schizophrenia
neurodevelopmental disorders. Biol. Psychiatry 68, Neuron 80, 624–632 (2013). and autism: both shared and disorder-specific
314–319 (2010). 10. Glynn, L. M., Wadhwa, P. D., Dunkel-Schetter, C., pathogenesis via perinatal inflammation? Pediatr. Res.
2. Curley, J. P., Jensen, C. L., Mashoodh, R. & Chicz-Demet, A. & Sandman, C. A. When stress 69, 26R–33R (2011).
Champagne, F. A. Social influences on neurobiology happens matters: effects of earthquake timing on This study examines the shared mechanistic link
and behavior: epigenetic effects during development. stress responsivity in pregnancy. Am. J. Obstet. between neurodevelopmental disorders that are
Psychoneuroendocrinology 36, 352–371 (2011). Gynecol. 184, 637–642 (2001). related to prenatal inflammation.
3. McCarthy, M. M. & Nugent, B. M. Epigenetic 11. Deverman, B. E. & Patterson, P. H. Cytokines and CNS 19. Morrison, K. E., Rodgers, A. B., Morgan, C. P. &
contributions to hormonally mediated sexual development. Neuron 64, 61–78 (2009). Bale, T. L. Epigenetic mechanisms in pubertal brain
differentiation of the brain. J. Neuroendocrinol. 12. Howerton, C. L. & Bale, T. L. Prenatal programing: at maturation. Neuroscience 264, 7–24 (2013).
25,1133–1140 (2013). the intersection of maternal stress and immune 20. Davis, E. P., Sandman, C. A., Buss, C., Wing, D. A. &
4. Hodes, G. E. Sex, stress, and epigenetics: regulation of activation. Horm. Behav. 62, 237–242 (2012). Head, K. Fetal glucocorticoid exposure is associated
behavior in animal models of mood disorders. Biol. 13. Brown, A. S. Epidemiologic studies of exposure to with preadolescent brain development. Biol.
Sex Differ. 4, 1 (2013). prenatal infection and risk of schizophrenia and Psychiatry 74, 647–655 (2013).
5. Bohacek, J., Gapp, K., Saab, B. J. & Mansuy, I. M. autism. Dev. Neurobiol. 72, 1272–1276 (2012). 21. Malter Cohen, M. et al. Early-life stress has persistent
Transgenerational epigenetic effects on brain 14. Brown, A. S. et al. Prenatal infection and cavum effects on amygdala function and development in mice
functions. Biol. Psychiatry 73, 313–320 (2013). septum pellucidum in adult schizophrenia. Schizophr. and humans. Proc. Natl Acad. Sci. USA 110,
6. Hales, C. N. & Barker, D. J. The thrifty phenotype Res. 108, 285–287 (2009). 18274–18278 (2013).
hypothesis. Br. Med. Bull. 60, 5–20 (2001). 15. Brown, A. S. et al. Prenatal exposure to maternal 22. Bale, T. L. Sex differences in prenatal epigenetic
In this paper, the thrifty phenotype hypothesis infection and executive dysfunction in adult programming of stress pathways. Stress 14, 348–356
describes how a mismatch between fetal schizophrenia. Am. J. Psychiatry 166, 683–690 (2011).
programming and the postnatal environment can (2009). 23. Susser, E., St Clair, D. & He, L. Latent effects of prenatal
result in disease risk. 16. Selevan, S. G., Kimmel, C. A. & Mendola, P. Identifying malnutrition on adult health: the example of schizophrenia.
7. Binder, E. B. & Nemeroff, C. B. The CRF system, stress, critical windows of exposure for children’s health. Ann. NY Acad. Sci. 1136, 185–192 (2008).
depression and anxiety-insights from human genetic Environ. Health Perspect. 108 (Suppl. 3), 451–455 24. Stoner, R. et al. Patches of disorganization in the
studies. Mol. Psychiatry 15, 574–588 (2010). (2000). neocortex of children with autism. N. Engl. J. Med.
8. Hackman, D. A., Farah, M. J. & Meaney, M. J. 17. Rapoport, J. L., Addington, A. M., Frangou, S. & 370, 1209–1219 (2014).
Socioeconomic status and the brain: mechanistic Psych, M. R. The neurodevelopmental model of 25. Avishai-Eliner, S., Brunson, K. L., Sandman, C. A. &
insights from human and animal research. Nature Rev. schizophrenia: update 2005. Mol. Psychiatry 10, Baram, T. Z. Stressed-out, or in (utero)? Trends
Neurosci. 11, 651–659 (2010). 434–449 (2005). Neurosci. 25, 518–524 (2002).

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | JUNE 2015 | 341

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

26. Wadhwa, P. D., Sandman, C. A. & Garite, T. J. The 48. Schneider, M. L., Moore, C. F., Kraemer, G. W., lasting consequences of early life stress. Endocrinology
neurobiology of stress in human pregnancy: Roberts, A. D. & DeJesus, O. T. The impact of prenatal 149, 4892–4900 (2008).
implications for prematurity and development of the stress, fetal alcohol exposure, or both on This novel and reproducible model shows how
fetal central nervous system. Prog. Brain Res. 133, development: perspectives from a primate model. disruptions in maternal care can manifest in
131–142 (2001). Psychoneuroendocrinology 27, 285–298 (2002). reprogramming of offspring stress pathways in the
27. Wadhwa, P. D., Sandman, C. A., Porto, M., 49. Kapoor, A. & Matthews, S. G. Short periods of brain.
Dunkel-Schetter, C. & Garite, T. J. The association prenatal stress affect growth, behaviour and 71. Ladd, C. O. et al. Long-term behavioral and
between prenatal stress and infant birth weight and hypothalamo–pituitary–adrenal axis activity in male neuroendocrine adaptations to adverse early
gestational age at birth: a prospective investigation. guinea pig offspring. J. Physiol. 566, 967–977 experience. Prog. Brain Res. 122, 81–103 (2000).
Am. J. Obstet. Gynecol. 169, 858–865 (1993). (2005). 72. Liu, D. et al. Maternal care, hippocampal
28. Khashan, A. S. et al. Higher risk of offspring 50. Mueller, B. R. & Bale, T. L. Early prenatal stress impact glucocorticoid receptors, and hypothalamic–pituitary–
schizophrenia following antenatal maternal exposure on coping strategies and learning performance is sex adrenal responses to stress. Science 277,
to severe adverse life events. Arch. Gen. Psychiatry dependent. Physiol. Behav. 91, 55–65 (2007). 1659–1662 (1997).
65, 146–152 (2008). 51. Lemaire, V., Koehl, M., Le Moal, M. & Abrous, D. N. 73. Meaney, M. J. Maternal care, gene expression, and
29. Atladottir, H. O. et al. Association of hospitalization for Prenatal stress produces learning deficits associated the transmission of individual differences in stress
infection in childhood with diagnosis of autism with an inhibition of neurogenesis in the hippocampus. reactivity across generations. Annu. Rev. Neurosci. 24,
spectrum disorders: a Danish cohort study. Arch. Proc. Natl Acad. Sci. USA 97, 11032–11037 (2000). 1161–1192 (2001).
Pediatr. Adolesc. Med. 164, 470–477 (2010). 52. Darnaudery, M. & Maccari, S. Epigenetic 74. Weaver, I. C. et al. Epigenetic programming by
30. Atladottir, H. O. et al. Maternal infection requiring programming of the stress response in male and maternal behavior. Nature Neurosci. 7, 847–854
hospitalization during pregnancy and autism spectrum female rats by prenatal restraint stress. Brain Res. (2004).
disorders. J. Autism Dev. Disord. 40, 1423–1430 Rev. 57, 571–585 (2008). This important paper was the first to establish
(2010). 53. Weinstock, M. Alterations induced by gestational epigenetic mechanisms in the developing brain that
31. Goines, P. E. et al. Increased midgestational IFNγ, IL‑4 stress in brain morphology and behaviour of the were altered by the quality of maternal care.
and IL‑5 in women bearing a child with autism: a case- offspring. Prog. Neurobiol. 65, 427–451 (2001). 75. Barha, C. K., Pawluski, J. L. & Galea, L. A. Maternal
control study. Mol. Autism 2, 13 (2011). 54. Coe, C. L. et al. Prenatal stress diminishes care affects male and female offspring working
32. Brown, A. S. & Susser, E. S. Prenatal nutritional neurogenesis in the dentate gyrus of juvenile rhesus memory and stress reactivity. Physiol. Behav. 92,
deficiency and risk of adult schizophrenia. Schizophr. monkeys. Biol. Psychiatry 54, 1025–1034 (2003). 939–950 (2007).
Bull. 34, 1054–1063 (2008). 55. Welberg, L. A., Seckl, J. R. & Holmes, M. C. Prenatal 76. Sanchez, M. M., Ladd, C. O. & Plotsky, P. M. Early
33. Xu, M. Q. et al. Prenatal malnutrition and adult glucocorticoid programming of brain corticosteroid adverse experience as a developmental risk factor for
schizophrenia: further evidence from the 1959–1961 receptors and corticotrophin-releasing hormone: later psychopathology: evidence from rodent and
Chinese famine. Schizophr. Bull. 35, 568–576 possible implications for behaviour. Neuroscience primate models. Dev. Psychopathol. 13, 419–449
(2009). 104, 71–79 (2001). (2001).
34. Ravelli, G. P., Stein, Z. A. & Susser, M. W. Obesity in 56. Welberg, L. A. & Seckl, J. R. Prenatal stress, 77. Caldji, C. et al. Maternal care during infancy regulates
young men after famine exposure in utero and early glucocorticoids and the programming of the brain. the development of neural systems mediating the
infancy. N. Engl. J. Med. 295, 349–353 (1976). J. Neuroendocrinol. 13, 113–128 (2001). expression of fearfulness in the rat. Proc. Natl Acad.
35. Heijmans, B. T. et al. Persistent epigenetic differences 57. Ward, I. L. Prenatal stress feminizes and Sci. USA 95, 5335–5340 (1998).
associated with prenatal exposure to famine in demasculinizes the behavior of males. Science 175, 78. Weaver, I. C., Diorio, J., Seckl, J. R., Szyf, M. &
humans. Proc. Natl Acad. Sci. USA 105, 82–84 (1972). Meaney, M. J. Early environmental regulation of
17046–17049 (2008). This is one of the first studies to determine the hippocampal glucocorticoid receptor gene expression:
36. Van Lieshout, R. J., Taylor, V. H. & Boyle, M. H. importance of sex-specific neurodevelopmental characterization of intracellular mediators and
Pre-pregnancy and pregnancy obesity and programming that can be disrupted by maternal potential genomic target sites. Ann. NY Acad. Sci.
neurodevelopmental outcomes in offspring: stress. 1024, 182–212 (2004).
a systematic review. Obes. Rev. 12, e548–e559 58. Ward, I. L. & Stehm, K. E. Prenatal stress feminizes 79. Korosi, A. & Baram, T. Z. The pathways from mother’s
(2011). juvenile play patterns in male rats. Physiol. Behav. 50, love to baby’s future. Front. Behav. Neurosci. 3, 27
37. Buss, C. et al. Impaired executive function mediates 601–605 (1991). (2009).
the association between maternal pre-pregnancy body 59. Baron-Cohen, S. et al. Elevated fetal steroidogenic 80. Graham, Y. P., Heim, C., Goodman, S. H., Miller, A. H.
mass index and child ADHD symptoms. PLoS ONE 7, activity in autism. Mol. Psychiatry 20, 369–376 & Nemeroff, C. B. The effects of neonatal stress on
e37758 (2012). (2015). brain development: implications for psychopathology.
38. Howerton, C. L., Morgan, C. P., Fischer, D. B. & 60. Moore, L. et al. Serum testosterone levels are related Dev. Psychopathol. 11, 545–565 (1999).
Bale, T. L. O‑GlcNAc transferase (OGT) as a placental to cognitive function in men with schizophrenia. 81. Lyons, D. M., Martel, F. L., Levine, S., Risch, N. J. &
biomarker of maternal stress and reprogramming of Psychoneuroendocrinology 38, 1717–1728 (2013). Schatzberg, A. F. Postnatal experiences and genetic
CNS gene transcription in development. Proc. Natl 61. Lombardo, M. V. et al. Fetal testosterone influences effects on squirrel monkey social affinities and
Acad. Sci. USA 110, 5169–5174 (2013). sexually dimorphic gray matter in the human brain. emotional distress. Horm. Behav. 36, 266–275
39. Mao, J. et al. Contrasting effects of different maternal J. Neurosci. 32, 674–680 (2012). (1999).
diets on sexually dimorphic gene expression in the 62. Gur, R. E. et al. A sexually dimorphic ratio of 82. Barr, C. S. et al. Sexual dichotomy of an interaction
murine placenta. Proc. Natl Acad. Sci. USA 107, orbitofrontal to amygdala volume is altered in between early adversity and the serotonin transporter
5557–5562 (2010). schizophrenia. Biol. Psychiatry 55, 512–517 (2004). gene promoter variant in rhesus macaques. Proc. Natl
40. Weaver, J. R., Susiarjo, M. & Bartolomei, M. S. This paper reports the dysmasculinization of limbic Acad. Sci. USA 101, 12358–12363 (2004).
Imprinting and epigenetic changes in the early brain regions in men with schizophrenia. 83. McGowan, P. O. et al. Epigenetic regulation of the
embryo. Mamm. Genome 20, 532–543 (2009). 63. Fenoglio, K. A. et al. Enduring, handling-evoked glucocorticoid receptor in human brain associates with
41. Mueller, B. R. & Bale, T. L. Impact of prenatal stress on enhancement of hippocampal memory function and childhood abuse. Nature Neurosci. 12, 342–348
long term body weight is dependent on timing and glucocorticoid receptor expression involves activation (2009).
maternal sensitivity. Physiol. Behav. 88, 605–614 of the corticotropin-releasing factor type 1 receptor. 84. Vukojevic, V. et al. Epigenetic modification of the
(2006). Endocrinology 146, 4090–4096 (2005). glucocorticoid receptor gene is linked to traumatic
42. Mueller, B. R. & Bale, T. L. Sex-specific programming 64. Fenoglio, K. A., Brunson, K. L. & Baram, T. Z. memory and post-traumatic stress disorder risk in
of offspring emotionality after stress early in Hippocampal neuroplasticity induced by early-life genocide survivors. J. Neurosci. 34, 10274–10284
pregnancy. J. Neurosci. 28, 9055–9065 (2008). stress: functional and molecular aspects. Front. (2014).
This study identifies sex-specific changes in the Neuroendocrinol. 27, 180–192 (2006). 85. Koshy, T. S., Sara, V. R., King, T. L. & Lazarus, L. The
placenta in response to early gestational stress 65. Ivy, A. S., Brunson, K. L., Sandman, C. & Baram, T. Z. influence of protein restriction imposed at various
that are associated with changes in offspring Dysfunctional nurturing behavior in rat dams with stages of pregnancy on fetal and placental
neurodevelopment. limited access to nesting material: a clinically relevant development. Growth 39, 497–506 (1975).
43. Brunton, P. J. & Russell, J. A. Prenatal social stress in model for early-life stress. Neuroscience 154, 86. Vucetic, Z. et al. Early life protein restriction alters
the rat programmes neuroendocrine and behavioural 1132–1142 (2008). dopamine circuitry. Neuroscience 168, 359–370
responses to stress in the adult offspring: sex-specific 66. Ivy, A. S. et al. Hippocampal dysfunction and cognitive (2010).
effects. J. Neuroendocrinol. 22, 258–271 (2010). impairments provoked by chronic early-life stress 87. Pankevich, D. E., Teegarden, S. L., Hedin, A. D.,
44. Brunton, P. J. & Russell, J. A. Allopregnanolone and involve excessive activation of CRH receptors. Jensen, C. L. & Bale, T. L. Caloric restriction
suppressed hypothalamo–pituitary–adrenal axis J. Neurosci. 30, 13005–13015 (2010). experience reprograms stress and orexigenic
stress responses in late pregnancy in the rat. Stress 67. Korosi, A. & Baram, T. Z. Plasticity of the stress pathways and promotes binge eating. J. Neurosci. 30,
14, 6–12 (2011). response early in life: mechanisms and significance. 16399–16407 (2010).
45. Weinstock, M. Gender differences in the effects of Dev. Psychobiol. 52, 661–670 (2010). 88. Teegarden, S. L. & Bale, T. L. Decreases in dietary
prenatal stress on brain development and behaviour. 68. Korosi, A. et al. Early-life experience reduces preference produce increased emotionality and risk for
Neurochem. Res. 32, 1730–1740 (2007). excitation to stress-responsive hypothalamic neurons dietary relapse. Biol. Psychiatry 61, 1021–1029
46. Kapoor, A., Kostaki, A., Janus, C. & Matthews, S. G. and reprograms the expression of corticotropin- (2007).
The effects of prenatal stress on learning in adult releasing hormone. J. Neurosci. 30, 703–713 (2010). 89. Teegarden, S. L. & Bale, T. L. Effects of stress on
offspring is dependent on the timing of the stressor. 69. McClelland, S., Korosi, A., Cope, J., Ivy, A. & dietary preference and intake are dependent on access
Behav. Brain Res. 197, 144–149 (2009). Baram, T. Z. Emerging roles of epigenetic mechanisms and stress sensitivity. Physiol. Behav. 93, 713–723
47. Kapoor, A., Leen, J. & Matthews, S. G. Molecular in the enduring effects of early-life stress and (2008).
regulation of the hypothalamic–pituitary–adrenal axis experience on learning and memory. Neurobiol. Learn. 90. Teegarden, S. L., Scott, A. N. & Bale, T. L. Early life
in adult male guinea pigs after prenatal stress at Mem. 96, 79–88 (2011). exposure to a high fat diet promotes long-term
different stages of gestation. J. Physiol. 586, 70. Rice, C. J., Sandman, C. A., Lenjavi, M. R. & changes in dietary preferences and central reward
4317–4326 (2008). Baram, T. Z. A novel mouse model for acute and long- signaling. Neuroscience 162, 924–932 (2009).

342 | JUNE 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

91. Will, M. J., Franzblau, E. B. & Kelley, A. E. Nucleus 113. Hodge, D. R. et al. Interleukin‑6 regulation of the This is a comprehensive examination, in adult male
accumbens μ‑opioids regulate intake of a high-fat diet human DNA methyltransferase (HDNMT) gene in mice, of epigenetic changes in germ cells that occur
via activation of a distributed brain network. human erythroleukemia cells. J. Biol. Chem. 276, in response to exposure to an odour cue in fear
J. Neurosci. 23, 2882–2888 (2003). 39508–39511 (2001). conditioning that is related to reprogramming of
92. Pecina, S., Cagniard, B., Berridge, K. C., Aldridge, J. W. 114. Howerton, C. L. & Bale, T. L. Targeted placental offspring brain and behaviour.
& Zhuang, X. Hyperdopaminergic mutant mice have deletion of OGT recapitulates the prenatal stress 134. Rodgers, A. B., Morgan, C. P., Bronson, S. L.,
higher “wanting” but not “liking” for sweet rewards. phenotype including hypothalamic mitochondrial Revello, S. & Bale, T. L. Paternal stress exposure alters
J. Neurosci. 23, 9395–9402 (2003). dysfunction. Proc. Natl Acad. Sci. USA 111, sperm microRNA content and reprograms offspring
93. Cottrell, E. C., Holmes, M. C., Livingstone, D. E., 9639–9644 (2014). HPA stress axis regulation. J. Neurosci. 33,
Kenyon, C. J. & Seckl, J. R. Reconciling the nutritional 115. Dunn, G. A., Morgan, C. P. & Bale, T. L. Sex-specificity 9003–9012 (2013).
and glucocorticoid hypotheses of fetal programming. in transgenerational epigenetic programming. Horm. This paper identifies important changes in specific
FASEB J. 26, 1866–1874 (2012). Behav. 59, 290–295 (2011). sperm mi­RNAs produced by chronic stress in adult
94. Lesage, J., Blondeau, B., Grino, M., Breant, B. & 116. Pembrey, M. E. et al. Sex-specific, male-line mice that correlate with programming of offspring
Dupouy, J. P. Maternal undernutrition during late transgenerational responses in humans. Eur. J. Hum. stress reactivity.
gestation induces fetal overexposure to glucocorticoids Genet. 14, 159–166 (2006). 135. Ng, S. F. et al. Chronic high-fat diet in fathers
and intrauterine growth retardation, and disturbs the 117. Kaati, G., Bygren, L. O. & Edvinsson, S. Cardiovascular programs β-cell dysfunction in female rat offspring.
hypothalamo–pituitary–adrenal axis in the newborn and diabetes mortality determined by nutrition during Nature 467, 963–966 (2010).
rat. Endocrinology 142, 1692–1702 (2001). parents’ and grandparents’ slow growth period. Eur. 136. Vassoler, F. M., White, S. L., Schmidt, H. D.,
95. Lingas, R., Dean, F. & Matthews, S. G. Maternal J. Hum. Genet. 10, 682–688 (2002). Sadri-Vakili, G. & Pierce, R. C. Epigenetic inheritance
nutrient restriction (48 h) modifies brain corticosteroid 118. Pembrey, M., Saffery, R. & Bygren, L. O. Human of a cocaine-resistance phenotype. Nature Neurosci.
receptor expression and endocrine function in the fetal transgenerational responses to early-life experience: 16, 42–47 (2013).
guinea pig. Brain Res. 846, 236–242 (1999). potential impact on development, health and 137. Carone, B. R. et al. Paternally induced
96. Cottrell, E. C. & Seckl, J. R. Prenatal stress, biomedical research. J. Med. Genet. 51, 563–572 transgenerational environmental reprogramming of
glucocorticoids and the programming of adult disease. (2014). metabolic gene expression in mammals. Cell 143,
Front. Behav. Neurosci. 3, 19 (2009). This is a comprehensive review that covers the field 1084–1096 (2010).
97. Seckl, J. R. & Holmes, M. C. Mechanisms of disease: of transgenerational epigenetics in relation to 138. Anway, M. D., Cupp, A. S., Uzumcu, M. &
glucocorticoids, their placental metabolism and fetal human health and disease. Skinner, M. K. Epigenetic transgenerational actions of
‘programming’ of adult pathophysiology. Nature Clin. 119. Morgan, C. P. & Bale, T. L. Early prenatal stress endocrine disruptors and male fertility. Science 308,
Pract. Endocrinol. Metab. 3, 479–488 (2007). epigenetically programs dysmasculinization in second- 1466–1469 (2005).
98. Welberg, L. A., Thrivikraman, K. V. & Plotsky, P. M. generation offspring via the paternal lineage. 139. Anderson, L. M. et al. Preconceptional fasting of
Chronic maternal stress inhibits the capacity to J. Neurosci. 31, 11748–11755 (2011). fathers alters serum glucose in offspring of mice.
up‑regulate placental 11β‑hydroxysteroid 120. Franklin, T. B. et al. Epigenetic transmission of the Nutrition 22, 327–331 (2006).
dehydrogenase type 2 activity. J. Endocrinol. 186, impact of early stress across generations. Biol. 140. Jenkins, T. G. & Carrell, D. T. The sperm epigenome
R7–R12 (2005). Psychiatry 68, 408–415 (2010). and potential implications for the developing embryo.
99. Grayson, B. E. et al. Changes in melanocortin 121. King, V. et al. Maternal obesity has little effect on Reproduction 143, 727–734 (2012).
expression and inflammatory pathways in fetal the immediate offspring but impacts on the next 141. van Os, J. & Selten, J. P. Prenatal exposure to
offspring of nonhuman primates fed a high-fat diet. generation. Endocrinology 154, 2514–2524 maternal stress and subsequent schizophrenia. The
Endocrinology 151, 1622–1632 (2010). (2013). May 1940 invasion of The Netherlands. Br.
100. Sullivan, E. L. et al. Chronic consumption of a high-fat 122. Dunn, G. A. & Bale, T. L. Maternal high-fat diet J. Psychiatry 172, 324–326 (1998).
diet during pregnancy causes perturbations in the promotes body length increases and insulin 142. Gerardin, P. et al. Depression during pregnancy: is the
serotonergic system and increased anxiety-like insensitivity in second-generation mice. Endocrinology developmental impact earlier in boys? A prospective
behavior in nonhuman primate offspring. J. Neurosci. 150, 4999–5009 (2009). case-control study. J. Clin. Psychiatry 72, 378–387
30, 3826–3830 (2010). 123. Dunn, G. A. & Bale, T. L. Maternal high-fat diet effects (2010).
101. Bilbo, S. D. & Tsang, V. Enduring consequences of on third-generation female body size via the paternal 143. Newschaffer, C. J. et al. The epidemiology of autism
maternal obesity for brain inflammation and behavior lineage. Endocrinology 152, 2228–2236 (2011). spectrum disorders. Annu. Rev. Publ. Health 28,
of offspring. FASEB J. 24, 2104–2115 (2010). 124. Jimenez-Chillaron, J. C. et al. Intergenerational 235–258 (2007).
102. Frias, A. E. et al. Maternal high-fat diet disturbs transmission of glucose intolerance and obesity by 144. Beversdorf, D. Q. et al. Timing of prenatal stressors
uteroplacental hemodynamics and increases the in utero undernutrition in mice. Diabetes 58, and autism. J. Autism Dev. Disord. 35, 471–478
frequency of stillbirth in a nonhuman primate model of 460–468 (2009). (2005).
excess nutrition. Endocrinology 152, 2456–2464 125. Thamotharan, M. et al. Transgenerational inheritance 145. Sandman, C. A., Glynn, L. M. & Davis, E. P. Is there a
(2011). of the insulin-resistant phenotype in embryo- viability–vulnerability tradeoff? Sex differences in fetal
103. Shen, Q. et al. The role of pro-inflammatory factors in transferred intrauterine growth-restricted adult female programming. J. Psychosomat. Res. 75, 327–335
mediating the effects on the fetus of prenatal rat offspring. Am. J. Physiol. Endocrinol. Metab. 292, (2013).
undernutrition: implications for schizophrenia. E1270–E1279 (2007). 146. Smith, S. E., Li, J., Garbett, K., Mirnics, K. &
Schizophr. Res. 99, 48–55 (2008). 126. Burdge, G. C. et al. Dietary protein restriction of Patterson, P. H. Maternal immune activation alters
104. Ilievski, V., Lu, S. J. & Hirsch, E. Activation of toll-like pregnant rats in the F0 generation induces altered fetal brain development through interleukin‑6.
receptors 2 or 3 and preterm delivery in the mouse. methylation of hepatic gene promoters in the adult J. Neurosci. 27, 10695–10702 (2007).
Reprod. Sci. 14, 315–320 (2007). male offspring in the F1 and F2 generations. Br. J. Nutr. 147. Morgan, C. P. & Bale, T. L. Sex differences in
105. Hsiao, E. Y. & Patterson, P. H. Placental regulation of 97, 435–439 (2007). microRNA regulation of gene expression: no smoke,
maternal–fetal interactions and brain development. 127. Boney, C. M., Verma, A., Tucker, R. & Vohr, B. R. just miRs. Biol. Sex Differ. 3, 22 (2012).
Dev. Neurobiol. 72,1317–1326 (2012). Metabolic syndrome in childhood: association with 148. Cowell, P. E., Kostianovsky, D. J., Gur, R. C.,
106. Malkova, N. V., Yu, C. Z., Hsiao, E. Y., Moore, M. J. & birth weight, maternal obesity, and gestational Turetsky, B. I. & Gur, R. E. Sex differences in
Patterson, P. H. Maternal immune activation yields diabetes mellitus. Pediatrics 115, e290–e296 neuroanatomical and clinical correlations in
offspring displaying mouse versions of the three core (2005). schizophrenia. Am. J. Psychiatry 153, 799–805
symptoms of autism. Brain Behav. Immun. 26, 128. McCurdy, C. E. et al. Maternal high-fat diet triggers (1996).
607–616 (2012). lipotoxicity in the fetal livers of nonhuman primates. 149. McCarthy, M. M. et al. The epigenetics of sex
107. Bilbo, S. D. & Schwarz, J. M. Early-life programming of J. Clin. Invest. 119, 323–335 (2009). differences in the brain. J. Neurosci. 29,
later-life brain and behavior: a critical role for the 129. Sullivan, E. L., Smith, M. S. & Grove, K. L. Perinatal 12815–12823 (2009).
immune system. Front. Behav. Neurosci. 3, 14 (2009). exposure to high-fat diet programs energy balance, 150. Nugent, B. M. & McCarthy, M. M. Epigenetic
108. Bronson, S. L. & Bale, T. L. Prenatal stress-induced metabolism and behavior in adulthood. underpinnings of developmental sex differences in the
increases in placental inflammation and offspring Neuroendocrinology 93, 1–8 (2011). brain. Neuroendocrinology 93, 150–158 (2011).
hyperactivity are male-specific and ameliorated by This paper examines the sex-specific 151. Westberry, J. M., Trout, A. L. & Wilson, M. E.
maternal antiinflammatory treatment. Endocrinology neurodevelopmental programming and resulting Epigenetic regulation of estrogen receptor-α gene
155, 2635–2646 (2014). increased anxiety in the non-human primate that expression in the mouse cortex during early postnatal
109. Hsiao, E. Y. & Patterson, P. H. Activation of the follows exposure to a maternal high-fat diet during development. Endocrinology 151, 731–740 (2010).
maternal immune system induces endocrine changes pregnancy. 152. Kurian, J. R., Olesen, K. M. & Auger, A. P. Sex
in the placenta via IL‑6. Brain Behav. Immun. 25, 130. Tamashiro, K. L., Terrillion, C. E., Hyun, J., Koenig, J. I. differences in epigenetic regulation of the estrogen
604–615 (2011). & Moran, T. H. Prenatal stress or high-fat diet receptor-α promoter within the developing preoptic
110. Lucassen, P. J. et al. Prenatal stress reduces postnatal increases susceptibility to diet-induced obesity in rat area. Endocrinology 151, 2297–2305 (2010).
neurogenesis in rats selectively bred for high, but not offspring. Diabetes 58, 1116–1125 (2009). 153. Murray, E. K., Hien, A., de Vries, G. J. & Forger, N. G.
low, anxiety: possible key role of placental 131. Shiell, A. W., Campbell, D. M., Hall, M. H. & Epigenetic control of sexual differentiation of the bed
11β‑hydroxysteroid dehydrogenase type 2. Eur. Barker, D. J. Diet in late pregnancy and glucose-insulin nucleus of the stria terminalis. Endocrinology 150,
J. Neurosci. 29, 97–103 (2009). metabolism of the offspring 40 years later. BJOG 107, 4241–4247 (2009).
111. Mairesse, J. et al. Maternal stress alters endocrine 890–895 (2000). 154. McCarthy, M. M. Molecular aspects of sexual
function of the feto-placental unit in rats. Am. 132. Dietz, D. M. & Nestler, E. J. From father to offspring: differentiation of the rodent brain.
J. Physiol. Endocrinol. Metab. 292, E1526–E1533 paternal transmission of depressive-like behaviors. Psychoneuroendocrinology 19, 415–427 (1994).
(2007). Neuropsychopharmacology 37, 311–312 (2012). 155. Bale, T. L. Stress sensitivity and the development of
112. Garbett, K. A., Hsiao, E. Y., Kalman, S., 133. Dias, B. G. & Ressler, K. J. Parental olfactory affective disorders. Horm. Behav. 50, 529–533 (2006).
Patterson, P. H. & Mirnics, K. Effects of maternal experience influences behavior and neural structure in 156. Bale, T. L. Neuroendocrine and immune influences on
immune activation on gene expression patterns in the subsequent generations. Naure Neurosci. 17, 89–96 the CNS: it’s a matter of sex. Neuron 64, 13–16
fetal brain. Transl. Psychiatry 2, e98 (2012). (2014). (2009).

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | JUNE 2015 | 343

© 2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

157. Cryan, J. F. & Dinan, T. G. Mind-altering 166. Zovkic, I. B., Guzman-Karlsson, M. C. & Sweatt, J. D. 175. O’Regan, D., Welberg, L. L., Holmes, M. C. &
microorganisms: the impact of the gut microbiota on Epigenetic regulation of memory formation and Seckl, J. R. Glucocorticoid programming of pituitary–
brain and behaviour. Nature Rev. Neurosci. 13, maintenance. Learn. Mem. 20, 61–74 (2013). adrenal function: mechanisms and physiological
701–712 (2012). 167. Zovkic, I. B. & Sweatt, J. D. Epigenetic mechanisms in consequences. Semin. Neonatol. 6, 319–329 (2001).
158. Dinan, T. G. & Cryan, J. F. Regulation of the stress learned fear: implications for PTSD. 176. Nyirenda, M. J., Welberg, L. A. & Seckl, J. R.
response by the gut microbiota: implications for Neuropsychopharmacology 38, 77–93 (2013). Programming hyperglycaemia in the rat through prenatal
psychoneuroendocrinology. Psychoneuroendocrinology 168. Mahgoub, M. & Monteggia, L. M. A role for histone exposure to glucocorticoids — fetal effect or maternal
37, 1369–1378 (2012). deacetylases in the cellular and behavioral influence? J. Endocrinol. 170, 653–660 (2001).
159. Clarke, G. et al. The microbiome–gut–brain axis mechanisms underlying learning and memory. Learn. 177. Heller, E. A. et al. Locus-specific epigenetic remodeling
during early life regulates the hippocampal Mem. 21, 564–568 (2014). controls addiction- and depression-related behaviors.
serotonergic system in a sex-dependent manner. Mol. 169. Adachi, M. & Monteggia, L. M. Decoding transcriptional Nature Neurosci. 17, 1720–1727 (2014).
Psychiatry 18, 666–673 (2013). repressor complexes in the adult central nervous 178. Brunner, A. M., Nanni, P. & Mansuy, I. M. Epigenetic
160. Borrelli, E., Nestler, E. J., Allis, C. D. & system. Neuropharmacology 80, 45–52 (2014). marking of sperm by post-translational modification of
Sassone-Corsi, P. Decoding the epigenetic language 170. Lester, B. M. et al. Behavioral epigenetics. Ann. NY histones and protamines. Epigenetics Chromatin 7, 2
of neuronal plasticity. Neuron 60, 961–974 Acad. Sci. 1226, 14–33 (2011). (2014).
(2008). 171. Morrison, K. E., Rodgers, A. B., Morgan, C. P. & 179. Bale, T. L. Lifetime stress experience:
161. Colvis, C. M. et al. Epigenetic mechanisms and gene Bale, T. L. Epigenetic mechanisms in pubertal brain transgenerational epigenetics and germ cell
networks in the nervous system. J. Neurosci. 25, maturation. Neuroscience 264, 17–24 (2014). programming. Dialogues Clin. Neurosci. 16, 297–305
10379–10389 (2005). 172. Waddington, C. H. Canalization of development and (2014).
162. Tsankova, N., Renthal, W., Kumar, A. & Nestler, E. J. genetic assimilation of acquired characters. Nature 180. Kotaja, N. & Sassone-Corsi, P. The chromatid body:
Epigenetic regulation in psychiatric disorders. Nature 183, 1654–1655 (1959). a germ-cell-specific RNA-processing centre. Nature
Rev. Neurosci. 8, 355–367 (2007). This paper laid the foundation for the field of epigenetic Rev. Mol. Cell Biol. 8, 85–90 (2007).
163. Day, J. J. et al. DNA methylation regulates associative programming by defining the ‘epigenetic landscape’.
reward learning. Nature Neurosci. 16, 1445–1452 173. Berger, S. L., Kouzarides, T., Shiekhattar, R. &
(2013). Shilatifard, A. An operational definition of epigenetics. Acknowledgements
164. Roth, T. L. & Sweatt, J. D. Regulation of chromatin Genes Dev. 23, 781–783 (2009). This work was supported by grants from the US National
structure in memory formation. Curr. Opin. Neurobiol. 174. Drake, A. J., Walker, B. R. & Seckl, J. R. Institutes of Health MH099910, MH104184, MH091258,
19, 336–342 (2009). Intergenerational consequences of fetal programming MH087597 and MH073030.
165. Sweatt, J. D. Experience-dependent epigenetic by in utero exposure to glucocorticoids in rats. Am.
modifications in the central nervous system. Biol. J. Physiol. Regul. Integr. Comp. Physiol. 288, Competing interests statement
Psychiatry 65, 191–197 (2009). R34–R38 (2005). The author declares no competing interests.

344 | JUNE 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


Copyright of Nature Reviews Neuroscience is the property of Nature Publishing Group and
its content may not be copied or emailed to multiple sites or posted to a listserv without the
copyright holder's express written permission. However, users may print, download, or email
articles for individual use.

You might also like